Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

Methods

A framework for ultra-low-input spatial tissue


proteomics
Graphical abstract Authors
Anuar Makhmut, Di Qin,
Sonja Fritzsche, Jose Nimo,
Janett König, Fabian Coscia

Correspondence
fabian.coscia@mdc-berlin.de

Highlights
d Detailed protocol and guidelines for ultra-low-input spatial
tissue proteomics

d Single-cell MS-based proteomics data obtained from archival


FFPE tissue

d MS signal allows to normalize across tissue types and sample


amounts

d Cytokines and transcriptional regulators quantified in tonsil


microregions

Makhmut et al., 2023, Cell Systems 14, 1002–1014


November 15, 2023 ª 2023 Elsevier Inc.
https://doi.org/10.1016/j.cels.2023.10.003 ll
ll

Methods
A framework for ultra-low-input
spatial tissue proteomics
Anuar Makhmut,1 Di Qin,1 Sonja Fritzsche,1 Jose Nimo,1 Janett König,1 and Fabian Coscia1,2,*
€ck-Center
1Max-Delbru for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany
2Lead contact
*Correspondence: fabian.coscia@mdc-berlin.de
https://doi.org/10.1016/j.cels.2023.10.003

SUMMARY

Spatial proteomics combining microscopy-based cell phenotyping with ultrasensitive mass-spectrometry-


based proteomics is an emerging and powerful concept to study cell function and heterogeneity in (patho)
physiology. However, optimized workflows that preserve morphological information for phenotype discovery
and maximize proteome coverage of few or even single cells from laser microdissected tissue are currently
lacking. Here, we report a robust and scalable workflow for the proteomic analysis of ultra-low-input archival
material. Benchmarking in murine liver resulted in up to 2,000 quantified proteins from single hepatocyte con-
tours and nearly 5,000 proteins from 50-cell regions. Applied to human tonsil, we profiled 146 microregions
including T and B lymphocyte niches and quantified cell-type-specific markers, cytokines, and transcription
factors. These data also highlighted proteome dynamics within activated germinal centers, illuminating sites
undergoing B cell proliferation and somatic hypermutation. This approach has broad implications in biomed-
icine, including early disease profiling and drug target and biomarker discovery. A record of this paper’s
transparent peer review process is included in the supplemental information.

INTRODUCTION requires integrated and multimodal pipelines. We recently intro-


duced deep visual proteomics (DVP),15 a new concept
Cells are the functional units of organs, which fulfill essential combining imaging-based (fluorescence or bright field) single-
physiological tasks in a spatially defined manner to maintain tis- cell phenotyping with unbiased MS-based proteomics for global
sue integrity.1 To analyze cell dynamics in space and time, proteome profiling with cell type and spatial resolution. To
powerful spatial genomics,2 epigenomics,3 transcriptomics,4–6 realize DVP, we developed an automated laser microdissection
and imaging-based proteomics7,8 methods have been devel- (LMD) workflow for the streamlined collection of nuclei, cells, or
oped to better understand cellular and molecular drivers of larger regions of interest (ROIs) directly into 96- or 384-well
health and disease states. As proteins are the biomolecules plates, thereby connecting whole-slide imaging and deep-
closest to the cellular phenotype determining cell identity and learning-based image analysis16 with ultrasensitive MS-based
function,9,10 spatial proteomics (SP) methods are particularly proteomics.17 This allowed the profiling of as little as 100 pheno-
promising for the study of human (patho)physiology. SP type-matched cells from archival tissue material while also pre-
methods with the single-cell resolution are dominated by tar- serving detailed cell type and spatial information. Further ad-
geted antibody-based methods such as imaging mass cytome- vances in sample preparation and MS acquisition recently
try11 (IMC) or multiplex immunofluorescence (mIF) imaging,8,12 pioneered the profiling of single-cell proteome heterogeneity in
where several dozen proteins can be analyzed at (sub)cellular cryosections of murine liver tissue,18 emphasizing the strong
resolution. However, although such methods are well suited spatial influence on the hepatocyte-specific proteome. Despite
for the large-scale screening of cellular phenotypes, they fall these promising proof-of-concept studies, a systematic evalua-
far short of capturing the actual complexity of the cellular tion and optimization of all experimental steps of IF microscopy-
proteome. It is estimated that single-cell types express more guided spatial tissue proteomics is still missing. In particular, the
than 10,000 unique proteins,9 which is complemented by analysis of few or even single cells of formalin-fixed and
millions of potential proteoforms, including splice variants, paraffin-embedded (FFPE) tissue collected by LMD has re-
post-translational modifications (PTMs), and protein sequence mained elusive and relies on optimized and robust ‘‘end-to-
variants.10,13,14 Liquid chromatography mass spectrometry end’’ protocols. The successful development of such integrated
(LC-MS)-based proteomics in contrast enables the study of pro- workflows could pave the way for a plethora of biomedical ap-
teomes at an unbiased (i.e., untargeted), quantitative, and sys- plications, including early disease proteome profiling studies
tem-wide level.9 The combination of both of these complemen- directly from archived patient material, where only a few cells
tary proteomic approaches is therefore highly desirable but can be present.

1002 Cell Systems 14, 1002–1014, November 15, 2023 ª 2023 Elsevier Inc.
ll
Methods

Here, we describe a scalable, robust, and easy-to-use proto- tion of the glass-type slides toward the label end of the slide
col optimized for the profiling of ultra-low-input archival tissue (Figure S1B), which can impede efficient LMD collection in
guided by whole-slide (IF) imaging. After benchmarking in murine this area if not prevented by the addition of glycerol.25 We
liver tissue, we applied our workflow to study the cell-type- alternatively recommend tissue mounting on the side of the
resolved proteome of B and T lymphocytes in different spatially slide, which is distant from the label. Frame slides on the con-
defined niches, guided by four-marker whole-slide IF imaging. trary are more suitable for diverse biological sample types (i.e.,
We finally provide detailed guidelines covering all aspects of tissue or cell culture) but are suboptimal for the collection of
our workflow (Methods S1), from antigen retrieval (AR) and stain- directly connected contours, for example, for gridded sam-
ing on LMD membrane slides, over manual or automated sample pling schemes as used for the nanoPOTS approach,20 as
processing, to MS data acquisition and analysis. this can ultimately lead to loss of overall membrane integrity.
Irrespective of the choice of AR (HIER or PIER), staining tech-
RESULTS nique (H&E or IF), or LMD slide type (PEN or PPS), proteomics
results from three different liver tissue amounts were highly
Optimizing LMD-based low-input FFPE tissue consistent (Figures 1C–1E). Tissue samples were processed
proteomics in 384-well low-binding plates using an MS-compatible,
Light microscopy is an integral part of SP workflows that organic solvent-based protocol, which included 60-min heat-
combine LMD with MS-based proteomics19–21 (Figure 1A). ing at 95 C for efficient formalin decrosslinking,26 sequential
Although hematoxylin and eosin (H&E) staining protocols are Lys-C and trypsin digestion and miniaturized solid-phase
well established for tissue sections mounted on specialized extraction. Compared with our previous DVP protocol, we
LMD membrane slides23 (Figure S1A), IF-based protocols, optimized our workflow for lower microliter volumes (1–2 mL)
which allow the sensitive detection of a higher number of to minimize peptide loss from surface adsorption while still be-
cell type and functional markers (typically 3–5 per imaging cy- ing pipette-able with standard laboratory equipment. At the
cle), require additional AR steps for epitope unmasking. The same time, this also allowed the integration of robotic sample
choice of the AR method can not only influence staining and preparation workflows (STAR Methods). In addition, we used
image quality but also impacts LMD membrane integrity, tis- an optimized 15-min active nano-LC gradient (Figures S1C–
sue collection efficiency, and potentially proteome coverage S1E) in combination with an optimal window design dia-PA-
of trace sample amounts. The evaluation and optimization of SEF27 method on a trapped-ion mobility spectrometry (TIMS)
AR and staining prior to MS is therefore important for ultra- mass spectrometer (Bruker timsTOF SCP) for improved sensi-
low input or even single-cell applications that strictly depend tivity and sample throughput. Using DIA-NN,28 we quantified
on highly efficient tissue collection and protein extraction, as more than 9,000 precursors and 1,700 unique proteins from
well as near-lossless sample preparation protocols. In other small tissue regions of approximately one to two hepatocytes
words, the most sensitive mass spectrometer can only be as (7,500 mm3, Figures 1C, S1F, and S1G). From 50-cell sam-
good as the upstream sample preparation workflow that de- ples (50,000 mm2, 5 mm thick) of H&E, HIER, and PIER-derived
livers these trace peptide amounts to the instrument. We samples, 4,000 proteins were consistently quantified with
therefore first evaluated different AR methods for their general excellent quantitative reproducibility (Pearson r = 0.98, Fig-
compatibility with whole-slide imaging on LMD slides, efficient ure 1D). Furthermore, we found a 95% overlap of protein iden-
LMD, and ultrasensitive MS-based proteomics. We tested two tifications across AR methods (Figure 1F), as well as nearly
common protocols based on heat-induced epitope retrieval identical cellular compartment proportions of the underlying
(HIER) or proteolytic (i.e., pepsin-induced epitope retrieval proteomes (Figure 1G), and consistent with a deep liver prote-
[PIER]) epitope retrieval and immunofluorescently stained mu- ome study of primary cells.22 We conclude that common FFPE
rine liver FFPE tissue with an antibody against a ubiquitously tissue preparation methods for fluorescence microscopy are
expressed plasma membrane marker (Na/K-ATPase). We fully compatible with ultra-low input MS-based proteomics.
chose murine liver as benchmarking tissue as hepatocytes However, depending on the concrete application, glass mem-
make up 60%–80% of the liver cell mass,24 which allowed brane or metal frame slides offer unique advantages and dis-
us to obtain consistent results from serial, homogeneous tis- advantages, which we summarized in Figure 1H to provide
sue slices, and repeated sample collections over the course general guidelines for LMD proteomics beginners. These
of our experiments. 5-mm-thick liver sections were mounted data also include input from two additional LMD expert labs
on glass membrane (polyethylene naphthalate [PEN] or poly- and together with our detailed protocol description (Methods
phenylene sulfide [PPS]) or metal frame (PPS) slides, de-paraf- S1; supplemental information), they are intended to support
finized, and subjected to two common AR protocols (STAR the selection of the right tissue preparation and sample collec-
Methods) prior to antibody staining, IF microscopy, LMD, tion strategy for diverse SP applications.
and proteomics (Figure 1B). We included H&E stains for com-
parison, which do not rely on additional AR after de-paraffini- A scalable FFPE tissue proteomics workflow allowing
zation, allowing us to directly investigate the impact of PIER single-cell analysis
and HIER on our proteomic results. PIER was fully compatible Having established that common AR and staining methods are
with both LMD slide types (glass or metal frame), facilitating fully compatible with LMD and ultra-low-input tissue proteomics,
efficient LMD, but generally came at the cost of higher back- we next assessed the scalability, robustness, and minimally
ground staining for the tested antibody, compared with HIER required sample amount of our workflow. Homogeneous areas
(Figure S1A). HIER occasionally resulted in membrane distor- of murine liver tissue were collected by LMD into 384-well

Cell Systems 14, 1002–1014, November 15, 2023 1003


ll
Methods

Figure 1. Optimizing laser microdissection-based low-input FFPE tissue proteomics


(A) Overview of the spatial tissue proteomics workflow.
(B) Tissue preparation strategy for laser microdissection-based proteomics benchmarking experiments using specialized LMD slides (metal frame or glass).
(C) Precursor and protein identifications from tissue samples processed with different staining and antigen retrieval methods. Areas of 1,562, 12,500, and
50,000 mm2 of a 5-mm thick section were laser microdissected. Averages are shown from quadruplicate measurements.
(D) Proteome correlations (Pearson’s r) of HIER, PIER, and H&E-based tissue samples.
(E) Boxplots showing coefficients of variations (CVs) of triplicate proteome measurements from the three antigen retrieval methods. Related to (D).
(F) Upset plot showing common and exclusive proteins for different antigen retrieval strategies based on 50,000 mm2 tissue samples.
(G) Protein identifications from major cellular compartments (‘‘cytosol,’’ ‘‘nucleus,’’ ‘‘plasma membrane,’’ and ‘‘extracellular region,’’ Gene Ontology Cellular
Component [GOCC]) from H&E, PIER, and HIER-treated mouse liver tissues. Percentages are the number of quantified proteins per compartment over all
quantified proteins in the corresponding sample. For comparison, a deep mouse liver proteome dataset was included22 based on non-fixed, primary cells.
(H) Summary of the laser-microdissection optimizations for low-input proteomics. Three labs assessed the applicability of glass and frame slides and rated each
category with moderate (+), good (++), and excellent (+++). The average score is shown from all three ratings. 1HIER can cause membrane distortion of glass-type
slides. 2Frame slides are more problematic for grid-based sampling schemes, the collection of many closely connected contours can lead to loss of overall
membrane integrity. 3Tested autostainers: Ventana (Roche) supported frame and glass slides, DAKO (Agilent) system supported glass slides. (A) and (B) created
with Biorender.

low-binding plates, ranging from single hepatocytes (600 mm2, ever, even single hepatocyte contours featured low median
5 mm thick) to approximately 100 cells (100,000 mm2, CVs of 20% for the 1,500–2,000 quantified proteins per contour
Figures 2A, 2B, S1F, and S1G). Proteomic results showed a (Figures 2E and S2A–S2C), which we and others previously only
linear increase in MS2 intensity, precursor, and protein identifi- achieved from many thousands of cells collected from FFPE tis-
cations from ‘‘low’’ to ‘‘high’’ tissue amounts, which, as ex- sue.30,31 It is noteworthy that CVs also included true biological
pected, was inversely correlated with the median coefficient of variation from known spatially defined hepatocyte heterogene-
variations (CV) of protein quantifications calculated from tripli- ity,18,32 letting us conclude that reproducible single-cell prote-
cate measurements of adjacent regions (Figures 2C–2E). How- ome analysis from FFPE tissue is achievable based on our

1004 Cell Systems 14, 1002–1014, November 15, 2023


ll
Methods

Figure 2. A scalable FFPE tissue proteomics workflow allowing single-cell analysis


(A) H&E-stained mouse liver tissue section (5 mm). Collected LMD tissue samples are shown from 600 up to 100,000 mm2. Single hepatocyte contour isolation
(average size 600 mm2) was guided by the immunofluorescence signal of the plasma membrane marker Na/K-ATPase and the DAPI signal. Larger regions of
interest (ROIs) were isolated based on circular contours of pre-defined sizes ranging from 1,562 to 10,000 mm2 (2–100 cells), respectively. Hepatocyte volumes of
4,000–6,000 mm329 were used for cell count estimations of circular ROIs. Scale bars, 400 mm.
(B) Tissue inspection after collection into a 384-well low-binding plate. Scale bars, 400 mm.
(C and D) Average MS2 quantities (C), number of identified precursors (D, left), and proteins (D, right) of the collected tissue samples are shown. For all amounts,
three replicates were collected and measured. The tissue optimum is highlighted in purple and single-cell contours in orange.
(E) Boxplots showing the CVs of protein quantification across different tissue areas. CVs were calculated from triplicates of non-log-transformed data. The
boxplots define the range of the data (whiskers), 25th and 75th percentiles (box), and medians (solid line).
(F and G) Impact of different lysis buffers on proteome coverage. Tissues of 7,500, 62,500, and 250,000 mm3 of a 5-mm thick mouse liver section were profiled
using three different methods based on organic solvent only (ACN), DDM only (no ACN), or combined (DDM and ACN) protocol. For all amounts, three replicates
were collected and measured.

(legend continued on next page)

Cell Systems 14, 1002–1014, November 15, 2023 1005


ll
Methods

workflow. We attribute the excellent quantitative reproducibility cyte-specific markers distributed over a dynamic range of
to the combination of optimized tissue preparation, low-volume approximately three orders of magnitude (Figures 2H and 2I).
sample processing, and highest-sensitivity MS acquisition and Proteins involved in ‘‘housekeeping’’ cell functions were quanti-
analysis using an optimal window dia-PASEF scheme (STAR fied at a similar depth of analysis as the 50-cell samples, despite
Methods). Precursor and protein identifications peaked for 50– the approximately 33-fold lower total MS2 signal, and, remark-
100 cell regions (25,000–50,000 mm2), where we quantified close ably, compared with the deep mouse liver study covering more
to 50,000 precursors and 5,000 proteins (Table S1). Interestingly, than 10,000 proteins22 (Figure 2J). For example, we quantified
further increasing sample amounts resulted in lower precursor 70 of 81 ribosomal proteins, 29 of 44 involved in fatty acid meta-
identifications and higher median CVs, indicative of sample over- bolism and 22 of 30 tricarboxylic acid (TCA) cycle-related pro-
loading (Figures 2D and 2E). Although this phenomenon could teins (Table S2). Interestingly, although housekeeping functions
possibly be balanced out using longer LC gradients and adjusted such as ribosomal proteins were more stably expressed, cell-
trypsin amounts, thereby further improving proteome coverage, metabolism-related proteins showed higher variation in the sin-
we note that our optimized 15-min active nanoflow gradient pro- gle-cell contours compared with the 50-cell contours, likely due
vides an excellent compromise between single-cell sensitivity, to proteome averaging (Figure S2I). This is in line with a recent
high proteome coverage for 50–100 cell samples, and reason- liver study revealing strong metabolic differences in single hepa-
able sample throughput of around 30–40 samples per day. Addi- tocytes along the liver zonation axis.18 For lower abundant path-
tionally, we tested the applicability of our protocol in combination ways, for example, insulin signaling or major histocompatibility
with the Bruker timsTOF Pro2, which shows a roughly 4- to 5-fold complex (MHC)-2 antigen presentation (12 vs. 107 and 25 vs.
lower total ion current (TIC) compared with the SCP instru- 92 detected proteins, respectively) (Figure 2J), a higher discor-
ment.17 For the lowest tissue amounts measured (7,500 mm3 dance in detected proteins was observed between the low and
samples, 1–2 hepatocytes29,33), close to 1,000 proteins could high tissue amounts, making the comprehensive single-cell anal-
still be quantified with high quantitative reproducibility and a ysis of these pathways only achievable with further improved
somewhat similar proteome coverage was observed for the workflow sensitivity, or alternatively, by pooling phenotype-
50-cell samples (Figures S2D–S2F). We next tested if we could matched cells, as we conceptualized recently.15 We conclude
further increase proteome coverage of single FFPE tissue con- that reproducible single-cell-based FFPE tissue profiling is
tours through the use of MS-compatible detergents. We as- achievable using optimized sample processing and ultrasensi-
sessed the integration of n-Dodecyl-b-D-maltoside (DDM), tive LC-MS workflows revealing important insights into cell
which was previously shown to improve proteome coverage of identity and function. The scalability of our workflow also enabled
low-input FFPE tissue, in particular when applied at high temper- the spatially resolved quantification of 5,000 proteins from
atures.34 Combining our acetonitrile (ACN) based protocol (10% 50-cell regions, capturing a substantial fraction of the cell-type-
final concentration) with DDM (0.1% final concentration), which specific proteome, thereby complementing single-cell-based
likewise included 60-min controlled heating at 95 C for efficient analyses.
formalin de-crosslinking, proteome coverage of the single-cell
samples improved by 104% and 50% for precursor and protein Optimizing sample input across tissue and cell types
identifications, respectively (Figures 2F and 2G). For 62,500 (12 Based on our tissue dilution experiment in the murine liver, we
cells) or 250,000 mm3 (50 cells) samples, proteome coverage empirically determined the optimal tissue amount for the highest
was more similar between protocols. As DDM is not removed proteome coverage of small tissue areas using a 15-min active
during peptide clean-up steps, its accumulation on the analytical nanoflow gradient combined with dia-PASEF on the Bruker tim-
column can compromise chromatographic performance over sTOF SCP. We next addressed how this liver optimum translated
time, which can be prevented by additional high organic solvent to other tissue and cell types and how the recorded MS readout
washing. Our organic-solvent-based protocol performed equally could be exploited to normalize sample loading from tissue to
well for 50-cell contours, thus offering an excellent and cleaner tissue or cell type to cell type. Such adjustments are of particular
alternative to the DDM/ACN combination for ‘‘higher’’ tissue importance for ultra-low sample amounts, which are not
amounts. amenable to peptide concentration measurements routinely
The unprecedented depth of our single FFPE hepatocyte con- used prior to MS bulk analysis. In addition, various factors can
tours, reproducibly quantifying up to 2,000 proteins depending affect the TIC derived from different tissue specimens, including
on tissue thickness (Figure S2C) and with high data complete- sample-related sources of variability such as tissue archival
ness (89% complete; Figure S2G), encouraged us to further time, which can affect the retrieval of lower abundant proteins,35
explore these single-cell tissue proteome data. Protein levels or biologically due to protein abundance differences across tis-
generally showed a high degree of concordance with higher sue and cell types.36 In any case, normalizing and adjusting sam-
loading amounts (Figure S2H) and included many known hepato- ple amounts is hence particularly important for low input tissue

(H) Dynamic range of protein abundance for 50-cell (250,000 mm3) contours. Proteins identified in single-cell contour samples are highlighted, as well as he-
patocyte-specific markers.22 A minimum of two quantified values per quadruplicate measurement was required for the single-contour samples.
(I) Histogram of log10 protein intensities obtained from 600 and 50,000 mm2 samples. Proteins identified in single cells cover 2–3 orders of magnitude from the top
abundant fraction of the liver proteome.
(J) Reactome and KEGG pathway coverage for single-cell and 50-cell samples. Values show the number of proteins quantified per pathway. For comparison, a
deep (>10,000 proteins) mouse liver dataset22 was included. A minimum of two quantified values from quadruplicate measurements was required for single cells
and three values from triplicates for the 50-cell samples.

1006 Cell Systems 14, 1002–1014, November 15, 2023


ll
Methods

Figure 3. Optimizing sample input across tissue and cell types


(A) Immunofluorescence whole-slide image of a 5-mm-thick tonsil tissue section stained for CD3 (T cells), CD19 (B cells), and DNA (DAPI). Scale bars, 500 mm.
(B) Magnifications of exemplary B and T cell enriched regions used for laser microdissection and proteomics profiling. Scale bars, 25 mm.

(legend continued on next page)

Cell Systems 14, 1002–1014, November 15, 2023 1007


ll
Methods

proteomics and should be carefully assessed. We hypothesized data output. To this end, we laser microdissected replicates of
that a simple normalization strategy using the total excised tis- small 60,000 mm3 samples (6,000 mm2 of a 10 mm slice) of a
sue volume (microdissected area 3 section thickness) across consecutive tonsil tissue section, such that the obtained inten-
specimens should be a poor estimator of the optimal sample sity was in the linear range of the mass spectrometric signal
amount. To test this, we analyzed a second tissue type, the hu- (>50,000 mm3 tissue; Figure 3D) and compared the recorded
man FFPE tonsil, which as a secondary lymphoid organ is impor- MS2 precursor quantity with our pre-determined optimum (MS
tant for the development of immune tolerance and adaptive im- signal = 6–8 3 1011). To minimize variability from different extra-
mune functions and comprises B and T cell subsets, as well as cellular matrix compositions, we focused on homogeneous tis-
other immune and non-immune-related cell types.37 In murine sue regions of high cellularity (Figure 3B), similar to the liver titra-
liver, 50–100 cell contours (125,000–250,000 mm3) yielded the tion experiment (Figure S1G). This way, we extrapolated that
highest proteome depth and lowest median protein CVs 450,000–753,000 mm3 would be the optimal sampling amount
(Figures 2C–2E and S3A–S3C). Analyzing the same tissue for this tonsil tissue, in excellent agreement with our measure-
amounts obtained from tonsil, focusing on T and B cell enriched ments (Figure 3C). We note that this simple normalization strat-
microregions after IF imaging (Figures 3A and 3B), revealed egy is strictly dependent on the employed LC-MS setup and
clearly different sampling optima (Figure 3C), despite a similar to- therefore requires empirical data to begin with but generally
tal number of quantified proteins per experiment (5,000–5,500 recommend to perform such ‘‘survey’’ experiments to pinpoint
proteins; Table S3). Based on the MS2 signal (sum of MS2 quan- the optimal tissue amount needed for a specific SP research
tities of all peaks) and total quantity (sum of MS2 quantities of question. In our example, murine liver served as highly consis-
identified precursors) retrieved from the DIA-NN output, we esti- tent reference tissue to predict the ideal tonsil amount; however,
mated that murine liver on average resulted in roughly 3-fold other tissue types should be equally suited for such normaliza-
(2.83) higher MS2 signal compared with tonsil (Figure 3D), likely tion. Moreover, this strategy can prevent TIMS overloading,
due to different protein abundances in these two organs (Fig- which can negatively affect proteome coverage and quantifica-
ure 3E). Notably, the MS2 signal derived from the liver reference tion. As a positive byproduct, the acquired raw files of the titra-
sample was consistent over the tested time frame of 6 months tion data can also be used to create project-specific refined
and independent of section thickness (Figure S3D). In other spectral libraries (STAR Methods), thereby drastically speeding
words, a 5-mm-thick contour of 25,000 mm2 resulted in an almost up subsequent DIA-NN searches.
identical MS2 signal as a 10-mm-thick contour of 12,500 mm2. As the size of the total dissected tissue area (or the number of
The optimal tissue amount for tonsil was 350,000–700,000 mm3 collected single-cell contours per sample), which determines the
instead, allowing us to quantify 35,000 precursors and obtained spatial resolution, is inversely correlated with proteome
5,000 proteins in the 15-min dia-PASEF measurement (Fig- coverage (Figures 2C and 2D), such survey measurements can
ure 3C), and beyond which no further increase in proteome depth also help to find a good balance between these two parameters
was apparent. In fact, we observed lower identification rates in the context of the specific research question. To demonstrate
beyond this saturation point, similar to our findings in liver this exemplarily, we further analyzed our tonsil proteome data,
(Figures 2C–2E), which we mostly attribute to increased tryptic which in total covered more than 5,000 proteins distributed
miscleavage (Figure S3E), as there was no sign of TIMS satura- over four orders of magnitude (Figure 3F). Projecting the different
tion for the tested tissue amounts. However, further increased tissue dilution measurements onto the measured dynamic range
peptide amounts could result in precursor-intensity-dependent of protein abundance showed that B or T cell-specific regions of
TIMS cell fragmentation, emphasizing the need to carefully only 8,750 mm2 (5-mm-thick section) were already sufficient to
assess and normalize sample loading. This prompted us to quantify many key players of immune cell signaling, cell-type-
test whether these tissue saturation points obtained from the specific markers, cytokines, and even transcription factors
liver and tonsil data could be exploited to predict the best tissue (e.g., STAT1, IRF3, IFNGR1, CD8A, CD19, Ki-67, LAG3, inter-
sampling amount from ‘‘single-shot’’ measurements alone. This leukin [IL]-16, and IL-18; Figures 3B, 3G, 3H, S3F, and S3G) at
simulates a scenario at the beginning of a spatial tissue prote- a spatial resolution of 70–100 mm (center-to-center; Figure 3A).
omics study, where a priori proteomics information for a new tis- Consequently, our pipeline should allow the analysis of spatially
sue type is lacking. The accurate prediction of the optimal tissue resolved proteomes of various B and T cell niches from regions
amount could hence save time and resources and maximize of as little as 4,000 mm2 (10-mm-thick section).

(C) Bar plots of MS2 intensities (left), precursors (middle) and proteins quantified (right) obtained from increasing amounts of tonsil tissue. For all amounts, six
replicates were collected and measured. Note, the tissue-specific sampling optimum was reached at 350,000–700,000 mm3, beyond which identifications
dropped again.
(D) Comparison of liver and tonsil tissue dilution data. MS2 quantities are plotted against increasing (log2 transformed) tissue amounts (volume in mm3). Based on
the empirically determined MS2 intensity optimum (MS2 quantity = 6–8E11 for liver and tonsil tissues), 2.8-fold more tonsil tissue was sampled to reach the same
MS2 quantity. For tonsil and liver tissue samples, data show averages from minimum six and five replicates per group, respectively.
(E) Cumulative protein intensities of liver and tonsil tissue proteomes ranked from the highest to the lowest abundant protein. Density plot shows protein intensity
distributions for both tissues. Top 20 proteins of each tissue are shown on the right.
(F) Dynamic range of protein abundance from different amounts of tonsil tissue.
(G) Histogram of protein intensities of the of 8,750 mm2 tonsil tissue sample (43,750 mm3 in volume). Proteins highlighted in pink belong to an RNA-seq-based
signature of tonsil germinal centers (GSE12845), including known immune and cell-type-specific markers.
(H) Unsupervised hierarchical clustering of small (6,000 mm2 3 10 mm) B cell, T cell, and mixed B/T cell regions, related to (B). Z scored protein levels indicate
upregulated (red) or downregulated (blue) proteins.

1008 Cell Systems 14, 1002–1014, November 15, 2023


ll
Methods

(legend on next page)

Cell Systems 14, 1002–1014, November 15, 2023 1009


ll
Methods

Spatially and cell-type-resolved proteomics of human proteome data. GCs showed clear signs of increased proliferation
tonsil tissue (e.g., Ki-67, PCNA) and DNA repair (e.g., TOP2A), indicative of
Encouraged by our tonsil titration data, which revealed that known active sites of B cell expansion and somatic hypermutation.40
immune cell regulators, cytokines, and transcription factors were T cell zones instead featured high levels of the STAT1 transcription
quantified from tissue regions of as little as 8,750 mm2 factor as well as T cell modulating cytokines such as IL-16 (Fig-
(43,750 mm3 in volume), we systematically investigated the impact ure 4F). Moreover, the global comparison of all T cells (n = 34) vs.
of the spatial location on the B and T cell specific proteomes. Hu- B cell-specific proteomes (mantle zone [MZ], n = 36) revealed
man tonsil represents a prime example of tissues that are orga- many bona fide B and T cell markers among the top regulated pro-
nized into distinct microanatomical compartments to fulfill diverse teins (e.g., CD22, CD3E, CD72, and CD5) and potentially many
biological functions critical for adaptive immunity. Following anti- other less-characterized ones (Figure 4G). We next focused on
gen encounter of naive B cells within the follicle, secondary (acti- different B cell niches to assess if our spatially resolved data
vated) follicles are rapidly formed as production centers of anti- captured known functional differences of spatially defined B cell
gen-specific B cells. Following activation, B cells undergo cycles zones. Naive (MZ) and activated (GC) B cells showed strong prote-
of maturation and selection within newly formed germinal centers ome differences (Figure 4H), indicative of their unique biological
(GCs), ultimately giving rise to highly antigen-specific antibody- functions. Although MZ B cells were characterized by higher
secreting plasma cells and memory B cells, the key players of hu- metabolic activity (e.g., TCA cycle and fructose and mannose
moral immune response.38 metabolism), senescence signatures, and phosphatidylinositol
We first mounted a 10-mm-thick tonsil section obtained from a signaling, activated GC B cells showed strong replication and
patient who underwent bilateral tonsillectomy on a metal frame DNA repair signatures (Figures 4H and 4I; Table S4), in line with
LMD slide and performed four-color IF whole-slide imaging to their known biological function. Encouraged by this, we next as-
detect B cells (CD19), T cells (CD3), epithelium (EP) (pan-CK), sessed whether our quantitative data even separated spatially
and DNA (DAPI) (Figures 4A and 4B). Using the open-source image defined GC sub-compartments of dark (sites undergoing active
analysis software QuPATH,39 we selected a total of 146 microre- B cell proliferation and somatic hypermutation) vs. light (site of B
gions for automated LMD (STAR Methods) and quantitative prote- cell selection) zones (Figures 4J and 4K). Clearly, dark-zone-
omics (Table S4). Based on the well-defined tissue architecture of derived B cells featured higher replication and DNA damage
the human tonsil (Figure 4A), we included small circular regions of response profiles compared with the light zone (Figures 4L and
4,000 mm2 isolated from primary and secondary B cell follicles, 4M). The higher expression of the T cell receptor subunit CD3D
including subregions of dark, light, and gray GC B cell niches, naive in light zones on the contrary illuminated the presence of T helper
mantel-zone-derived B cells, various interfollicular T cell zones, cells, important for B cell selection.40 We confirmed this finding by
and squamous cell EP (Figures 4A–4C). On average, we quantified assessing our imaging data, which indeed revealed significantly
1,952 proteins per sample (Figure S4C) and 3,334 proteins in total. higher CD3 signals in the light GC regions, which was not the
After data filtering and missing value imputation (STAR Methods), case for the B cell marker CD19 (Figure S4D).
proteomes clearly separated by microanatomical regions domi- In summary, our data delineated how the robust microscopy-
nated by the distinct cell types (Figure 4D). Known cell type guided ultra-low-input tissue proteomics workflow introduced
markers such as CD3D (T cell marker), CD19 (B cell marker), and here can be applied to study cell type and spatially resolved pro-
CDH1 (E-Cadherin, epithelial marker) were highest in the expected teomes in health and disease based on readily accessible
sample groups (Figure 4E), confirming the high specificity of our archival FFPE specimens.

Figure 4. Spatially and cell-type-resolved proteomics of human tonsil tissue


(A) Immunofluorescence whole-slide image of a 10-mm thick tonsil tissue section stained for CD3 (T cells), CD19 (B cells), pan-CK (epithelium), and DNA (DAPI).
Scale bars, 250 mm.
(B) Magnifications of the exemplary epithelium (EP), germinal center (GC), mantle zone (MZ), and T cell enriched regions used for laser microdissection and
proteomic profiling. Scale bars, 20 mm.
(C) Sample collection strategy and the total number of samples for each tissue region.
(D) 3D principal-component analysis (PCA) of 146 samples based on 2,235 protein groups after data filtering and imputation.
(E and F) Log2-protein levels of cell-type-specific and functional markers quantified in different tissue regions. Black dots indicate average values for each group.
Asterisks indicate ANOVA p values of <0.001 after data imputation.
(G) Volcano plot of the pairwise proteomic comparison between the B cell zone (mantle zone) and T cell zone. Cell type-specific markers are highlighted in green
and turquoise (two-sided t test, false discovery rate [FDR] < 0.05).
(H) Volcano plot of the pairwise proteomic comparison between mantle zone and germinal center samples. Cell type-specific markers are highlighted in green and
blue (two-sided t test, FDR < 0.05).
(I) Pathway enrichment analysis (Reactome and KEGG) based on t test difference between mantel zone and germinal center samples. Selected pathways with a
Benjamin-Hochberg FDR < 0.05 are shown.
(J) ROIs used for proteomic profiling of a secondary follicle region. Dark (red), gray (orange), and light (yellow) germinal center zones were selected for proteomic
profiling. Mantel zone regions (light gray) are shown additionally. Scale bars, 50 mm.
(K) PCA of dark, gray, and light zone proteomes. Point concentration ellipses are shown for each group with a 95% confidence.
(L) Boxplots of relative protein levels (Z score) for selected markers. The boxplots define the range of the data (whiskers), 25th and 75th percentiles (box), and
medians (solid line). Asterisks indicate two-sided t test p values (dark vs. light zone) of p < 0.001.
(M) Unsupervised hierarchical clustering of ANOVA significant proteins (permutation-based FDR < 0.05) from dark, gray, and light zone samples. Related to
(J) and (K). Heatmap shows relative protein levels (Z score) of upregulated (yellow) and downregulated proteins (black). *KRT78 is marked as potential
contaminant.

1010 Cell Systems 14, 1002–1014, November 15, 2023


ll
Methods

DISCUSSION normalization strategy using the extracted MS2 signal directly


from the search results to find the optimal tissue amount for liter-
Spatial tissue proteomics connecting microscopy-based cell ally any low-input spatial tissue proteomics experiment. This
phenotyping with LMD guided MS-based proteomics is an strategy can also be applied to normalize qualitative sample dif-
emerging discovery concept for the study of cell function and ferences, which are often observed for archival specimens due
heterogeneity in health and disease. Our group recently co- to, for example, varying archival times.31 The importance of
developed deep visual proteomics, an approach that combines this can be illustrated through the eyes of a pathologist. For
high-parametric imaging and machine-learning-based single- the histomolecular analysis of cancer progression states, sam-
cell phenotyping to guide precise tissue sampling for ultrasensi- ples are typically distributed over several FFPE tissue blocks
tive LC-MS analysis. This enabled the profiling of as little as 100 (for example pre-cancer, primary tumor, and metastasis), poten-
tissue cells per sample to a depth of 3,000–5,000 proteins, tially collected over many years. Survey experiments, as outlined
dependent on the tissue and cell type of interest. However, the here using the example of human tonsil tissue, could guide the
flexible and highly modular design of the DVP pipeline, enabling best sampling strategy for deep and reproducible LMD-assisted
the profiling of single or few cells on one hand, or hundreds of proteomics.
phenotype-matched cells for deeper proteome interrogation However, although the latest generation MS instruments
(i.e., 5,000 proteins or more) on the other hand, also necessitates feature excellent sensitivity when combined with optimized sam-
carefully designed tissue benchmarking experiments and ple preparation workflows, such as the one introduced here, MS
detailed guidelines to extract most information for diverse throughput is still a major bottleneck for tissue sections that are
biomedical applications. With this in mind, we here introduce, often larger than 1 cm2. Isobaric or non-isobaric multiplexing
benchmark, and apply an optimized end-to-end workflow, start- strategies generally offer good alternatives to label-free based
ing from AR comparisons for immunostaining and microscopy, methods further increasing sample throughput,44–46 but they
over guidelines for best-practice tissue collection by LMD, to- are still not sufficient to deal with this tremendous analytical
ward an optimal nanoflow dia-PASEF LC-MS scheme for high- bottleneck. One can estimate that for the profiling of one 1 3
est-sensitivity MS-based proteomics. For the first time, we pro- 1 cm tissue section, sampled by non-overlapping 50 3 50 mm
vide evidence that robust ultra-low-input proteomics (few- or squares, amounts, which we here show in liver and tonsil tissues
even single-excised cells) from FFPE tissue slices is achievable to be amenable to the reproducible quantification of 2,000–3,000
and fully compatible with conventional AR and four-marker IF im- proteins, 40,000 measurements would be required for gridded
aging protocols, paving the way for higher-plex IF combinations whole-slide sampling schemes. This is far beyond the reach of
in the near future. At the same time, our flexible 384-well design current LC-MS setups, which typically analyze 20–50 tissue pro-
makes the protocol easily adaptable to robotic workflows for teomes per day. Instead, the integration of whole-slide IF imag-
further protocol automation. We find that low microliter volumes ing for detailed cell and cellular neighborhood phenotyping al-
(2 mL) for sample preparation are sufficient to achieve true sin- lows us to prioritize cells and ROIs subjected to global
gle-cell sensitivity, which makes this workflow principally acces- proteome analysis, thereby offering a powerful, cost-effective,
sible to any laboratory. On the Bruker timsTOF SCP, this allowed and accessible spatial profiling strategy. We illustrate this in
us to quantify up to 2,000 proteins from single hepatocyte con- tonsil tissue, where we use four-marker whole-slide IF imaging
tours and around 5,000 proteins from 50-cell regions, demon- to guide the sampling of over 140 microregions per single batch,
strating excellent workflow scalability. On the timsTOF Pro2 in- covering naive and activated B cell niches, interfollicular T cell
strument, this translated to nearly 1,000 high-confidence zones and squamous cell EP. From only 63 3 63 mm regions
proteins from 1 to 2 hepatocytes (1,500 mm2 regions) and a (4,000 mm2 regions), we quantified spatially resolved proteomes
somewhat similar proteome depth for the 50-cell samples, of activated GC niches, illuminating sites of clonal B cell expan-
further emphasizing the broad applicability of our protocol. The sion and somatic hypermutation. Intriguingly, based on these
data from single-excised hepatocytes also revealed that a 1,500–3,000 protein measurements per microregion, we quanti-
25-mm spatial resolution is principally achievable for tissue fied key players of immune cell function including cytokines and
types such as liver, on par with the spatial resolution of state- transcriptional regulators, emphasizing the power of our ‘‘biolog-
of-the-art spatial transcriptomics.41,42 These data also show ical fractionation’’ strategy to dig deep into the cell type and
that highly abundant housekeeping proteins, such as ribosomal spatially resolved tissue proteome.
proteins, are quantifiable at a depth and precision similar to In conclusion, we here provide a scalable and optimized
much higher sampling amounts, for example, from large-scale framework for MS-based spatial tissue proteomics of ultra-
bulk measurements. Our data also show that many metabolic low-input archival specimens combining high-content imaging,
pathways (e.g., TCA cycle or fatty acid metabolism) are likewise LMD, and ultrasensitive MS.
amenable to single-cell-based proteomic analysis, as also
shown recently.18 Notably, housekeeping related proteins are STAR+METHODS
often poorly correlated to mRNA abundances,10,43 making these
pathways particularly attractive for early applications of single- Detailed methods are provided in the online version of this paper
cell-based tissue proteomics. and include the following:
We also provide guidance on how to empirically determine the
optimal tissue amount to achieve the highest proteome coverage d KEY RESOURCES TABLE
while avoiding instrument overloading, which is particularly perti- d RESOURCE AVAILABILITY
nent for single-cell sensitivity MS setups. We introduce a simple B Lead contact

Cell Systems 14, 1002–1014, November 15, 2023 1011


ll
Methods
B Materials availability sequencing. Cell 172, 205–217.e12. https://doi.org/10.1016/j.cell.2017.
B Data and code availability 12.007.
d EXPERIMENTAL MODEL AND SUBJECT DETAILS 3. Deng, Y., Bartosovic, M., Kukanja, P., Zhang, D., Liu, Y., Su, G., Enninful,
B Mouse experiments and organ harvesting A., Bai, Z., Castelo-Branco, G., and Fan, R. (2022). Spatial-CUT&Tag:
B Human tissue samples spatially resolved chromatin modification profiling at the cellular level.
Science 375, 681–686. https://doi.org/10.1126/science.abg7216.
d METHOD DETAILS
4. Marx, V. (2021). Method of the Year: spatially resolved transcriptomics.
B Hematoxylin-eosin staining
Nat. Methods 18, 9–14. https://doi.org/10.1038/s41592-020-01033-y.
B Immunofluorescence staining
5. Longo, S.K., Guo, M.G., Ji, A.L., and Khavari, P.A. (2021). Integrating single-
B High-resolution microscopy
cell and spatial transcriptomics to elucidate intercellular tissue dynamics. Nat.
B Image analysis and contour export for laser microdis-
Rev. Genet. 22, 627–644. https://doi.org/10.1038/s41576-021-00370-8.
section
6. He, S., Bhatt, R., Brown, C., Brown, E.A., Buhr, D.L., Chantranuvatana, K.,
B Laser microdissection Danaher, P., Dunaway, D., Garrison, R.G., Geiss, G., et al. (2022). High-
B Sample preparation for LC-MS analysis plex multiomic analysis in FFPE at subcellular level by spatial. Mol.
B Peptide clean-up with C-18 tips Imaging 40, 1794–1806. https://doi.org/10.1101/2021.11.03.467020.
B Liquid chromatography–mass spectrometry (LC – MS) 7. Baharlou, H., Canete, N.P., Cunningham, A.L., Harman, A.N., and Patrick,
analysis E. (2019). Mass cytometry imaging for the study of human diseases—ap-
B Proteomics raw data analysis plications and data analysis strategies. Front. Immunol. 10, 2657. https://
d QUANTIFICATION AND STATISTICAL ANALYSIS doi.org/10.3389/fimmu.2019.02657.
8. Schu €rch, C.M., Bhate, S.S., Barlow, G.L., Phillips, D.J., Noti, L., Zlobec, I.,
Chu, P., Black, S., Demeter, J., McIlwain, D.R., et al. (2020). Coordinated
SUPPLEMENTAL INFORMATION
cellular neighborhoods orchestrate antitumoral immunity at the colorectal
cancer invasive front. Cell 182, 1341–1359.e19. https://doi.org/10.1016/j.
Supplemental information can be found online at https://doi.org/10.1016/j.
cell.2020.07.005.
cels.2023.10.003.
9. Aebersold, R., and Mann, M. (2016). Mass-spectrometric exploration of
ACKNOWLEDGMENTS proteome structure and function. Nature 537, 347–355. https://doi.org/
10.1038/nature19949.
We would like to thank our colleagues at the Max Delbru €ck Center (MDC) for 10. Buccitelli, C., and Selbach, M. (2020). mRNAs, proteins and the emerging
their support and fruitful discussions. In particular, we thank Ulrike Stein for principles of gene expression control. Nat. Rev. Genet. 21, 630–644.
her support to perform the mouse liver experiments. We thank Philipp Mertins https://doi.org/10.1038/s41576-020-0258-4.
and Simon Haas for their critical feedback on the manuscript and Christian 11. Jackson, H.W., Fischer, J.R., Zanotelli, V.R.T., Ali, H.R., Mechera, R.,
Sommer for MS support. We also thank Andreas Mund (Center for Protein Soysal, S.D., Moch, H., Muenst, S., Varga, Z., Weber, W.P., et al. (2020).
Research, University of Copenhagen) and Lisa Schweizer (MPI of Biochem- The single-cell pathology landscape of breast cancer. Nature 578,
istry, Munich) for their input and feedback on the LMD membrane compari- 615–620. https://doi.org/10.1038/s41586-019-1876-x.
sons. We thank Simon Schallenberg (Charité Pathology, Berlin) for his help
12. Lin, J.R., Izar, B., Wang, S., Yapp, C., Mei, S., Shah, P.M., Santagata, S.,
with the tonsil tissue experiments. Furthermore, we acknowledge the MDC
and Sorger, P.K. (2018). Highly multiplexed immunofluorescence imaging
technology platforms ‘‘Proteomics’’ and ‘‘Advanced Light Microscopy’’ for
of human tissues and tumors using t-CyCIF and conventional optical mi-
their great support. All authors acknowledge support by the Federal Ministry
croscopes. eLife 7, 1–46. https://doi.org/10.7554/eLife.31657.
of Education and Research (BMBF), as part of the National Research Initiatives
for Mass Spectrometry in Systems Medicine, under grant agreement No. 13. Sharma, K., D’Souza, R.C.J., Tyanova, S., Schaab, C., Wisniewski, J.R.,
161L0222. Cox, J., and Mann, M. (2014). Ultradeep human phosphoproteome reveals
a distinct regulatory nature of Tyr and ser/Thr-based signaling. Cell Rep. 8,
1583–1594. https://doi.org/10.1016/j.celrep.2014.07.036.
AUTHOR CONTRIBUTIONS
14. Sinitcyn, P., Richards, A.L., Weatheritt, R.J., Brademan, D.R., Marx, H.,
Conceptualization, A.M. and F.C.; methodology, A.M., D.Q., J.K., J.N., and Shishkova, E., Meyer, J.G., Hebert, A.S., Westphall, M.S., Blencowe,
F.C.; experiments, A.M., D.Q., S.F., J.K., and F.C.; data analysis, A.M. and B.J., et al. (2023). Global detection of human variants and isoforms by
F.C.; figures, A.M. and F.C.; supervision, F.C.; funding acquisition, F.C.; writing deep proteome sequencing. Nat. Biotechnol. https://doi.org/10.1038/
the original draft, F.C. All the authors reviewed and edited the manuscript. s41587-023-01714-x.
15. Mund, A., Coscia, F., Kriston, A., Hollandi, R., Kovács, F., Brunner, A.-D.,
DECLARATION OF INTERESTS Migh, E., Schweizer, L., Santos, A., Bzorek, M., et al. (2022). Deep Visual
proteomics defines single-cell identity and heterogeneity. Nat. Biotechnol.
The authors declare no competing interests.
40, 1231–1240. https://doi.org/10.1038/s41587-022-01302-5.

Received: June 1, 2023 16. Hollandi, R., Szkalisity, A., Toth, T., Tasnadi, E., Molnar, C., Mathe, B.,
Revised: August 3, 2023 Grexa, I., Molnar, J., Balind, A., Gorbe, M., et al. (2020). nucleAIzer: A
Accepted: October 6, 2023 parameter-free deep learning framework for nucleus segmentation using
Published: October 30, 2023 image style transfer. Cell Syst. 10, 453–458.e6. https://doi.org/10.1016/j.
cels.2020.04.003.
REFERENCES 17. Brunner, A.D., Thielert, M., Vasilopoulou, C., Ammar, C., Coscia, F., Mund,
A., Hoerning, O.B., Bache, N., Apalategui, A., Lubeck, M., et al. (2022).
1. Palla, G., Fischer, D.S., Regev, A., and Theis, F.J. (2022). Spatial compo- Ultra-high sensitivity mass spectrometry quantifies single-cell proteome
nents of molecular tissue biology. Nat. Biotechnol. 40, 308–318. https:// changes upon perturbation. Mol. Syst. Biol. 18, e10798. https://doi.org/
doi.org/10.1038/s41587-021-01182-1. 10.15252/msb.202110798.
2. Casasent, A.K., Schalck, A., Gao, R., Sei, E., Long, A., Pangburn, W., 18. Rosenberger, F.A., Thielert, M., Strauss, M.T., Ammar, C., Ma €dler, C.,
Casasent, T., Meric-Bernstam, F., Edgerton, M.E., and Navin, N.E. (2018). Schweizer, L., Metousis, A., Skowronek, P., Wahle, M., Semenova, A.,
Multiclonal invasion in breast tumors identified by topographic single cell et al. (2023). Spatial single-cell mass spectrometry defines zonation of the

1012 Cell Systems 14, 1002–1014, November 15, 2023


ll
Methods
hepatocyte proteome author list. Nat Methods 20, 1530–1536. https://doi. 33. Milo, R., Jorgensen, P., Moran, U., Weber, G., and Springer, M. (2010).
org/10.1038/s41592-023-02007. BioNumbers—the database of key numbers in molecular and cell biology.
Nucleic Acids Res. 38, D750–D753. https://doi.org/10.1093/NAR/GKP889.
19. Wisniewski, J.R., Ostasiewicz, P., and Mann, M. (2011). High recovery
FASP applied to the proteomic analysis of microdissected formalin fixed 34. Nwosu, A.J., Misal, S.A., Truong, T., Carson, R.H., Webber, K.G.I., Axtell,
paraffin embedded cancer tissues retrieves known colon cancer markers. N.B., Liang, Y., Johnston, S.M., Virgin, K.L., Smith, E.G., et al. (2022). In-
J. Proteome Res. 10, 3040–3049. https://doi.org/10.1021/pr200019m. Depth mass spectrometry-Based proteomics of formalin-Fixed, Paraffin-
Embedded Tissues with a Spatial Resolution of 50–200 mm. J. Proteome
20. Piehowski, P.D., Zhu, Y., Bramer, L.M., Stratton, K.G., Zhao, R., Orton,
Res. 21, 2237–2245. https://doi.org/10.1021/acs.jproteome.2c00409.
D.J., Moore, R.J., Yuan, J., Mitchell, H.D., Gao, Y., et al. (2020).
Automated mass spectrometry imaging of over 2000 proteins from tissue 35. Balgley, B.M., Guo, T., Zhao, K., Fang, X., Tavassoli, F.A., and Lee, C.S.
sections at 100-mm spatial resolution. Nat. Commun. 11, 8. https://doi.org/ (2009). Evaluation of archival time on shotgun proteomics of formalin-fixed
10.1038/s41467-019-13858-z. and paraffin-embedded tissues. J. Proteome Res. 8, 917–925. https://doi.
org/10.1021/pr800503u.
21. Eckert, M.A., Coscia, F., Chryplewicz, A., Chang, J.W., Hernandez, K.M.,
, I., et al. (2019).
Pan, S., Tienda, S.M., Nahotko, D.A., Li, G., Blazenovic 36. Wang, D., Eraslan, B., Wieland, T., Hallström, B., Hopf, T., Zolg, D.P.,
Proteomics reveals NNMT as a master metabolic regulator of cancer- Zecha, J., Asplund, A., Li, L.H., Meng, C., et al. (2019). A deep proteome
associated fibroblasts. Nature 569, 723–728. https://doi.org/10.1038/ and transcriptome abundance atlas of 29 healthy human tissues. Mol.
s41586-019-1173-8. Syst. Biol. 15, e8503. https://doi.org/10.15252/MSB.20188503.
22. Azimifar, S.B., Nagaraj, N., Cox, J., and Mann, M. (2014). Cell-type- 37. Massoni-Badosa, R., Soler-Vila, P., Aguilar-Fernández, S., Nieto, J.C.,
resolved quantitative proteomics of murine liver. Cell Metab. 20, 1076– Elosua-Bayes, M., Marchese, D., Kulis, M., Vilas-Zornoza, A., Bu €hler,
1087. https://doi.org/10.1016/j.cmet.2014.11.002. M.M., Rashmi, S., et al. (2022). An atlas of cells in the human tonsil.
https://doi.org/10.1101/2022.06.24.497299.
23. Emmert-Buck, M.R., Bonner, R.F., Smith, P.D., Chuaqui, R.F., Zhuang, Z.,
Goldstein, S.R., Weiss, R.A., and Liotta, L.A. (1996). Laser capture micro- 38. De Silva, N.S., and Klein, U. (2015). Dynamics of B cells in germinal cen-
dissection. Science 274, 998–1001. https://doi.org/10.1126/science.274. tres. Nat. Rev. Immunol. 15, 137–148. https://doi.org/10.1038/nri3804.
5289.998. 39. Bankhead, P., Loughrey, M.B., Fernández, J.A., Dombrowski, Y., McArt,
24. Ben-Moshe, S., and Itzkovitz, S. (2019). Spatial heterogeneity in the D.G., Dunne, P.D., McQuaid, S., Gray, R.T., Murray, L.J., Coleman, H.G.,
mammalian liver. Nat. Rev. Gastroenterol. Hepatol. 16, 395–410. https:// et al. (2017). QuPath: open source software for digital pathology image anal-
doi.org/10.1038/s41575-019-0134-x. ysis. Sci. Rep. 7, 16878. https://doi.org/10.1038/s41598-017-17204-5.

25. Nordmann, T.M., Schweizer, L., Metousis, A., and Thielert, M. (2023). 40. Victora, G.D., and Nussenzweig, M.C. (2012). Germinal centers. Annu.
Rahbek-gjerdrum, L.M., Stadler, P, and Bzorek, M. A standardized and Rev. Immunol. 30, 429–457. https://doi.org/10.1146/annurev-immunol-
reproducible workflow for membrane glass slides in routine histology 020711-075032.
and spatial proteomics. https://doi.org/10.1101/2023.02.20.529255. 41. Lewis, S.M., Asselin-Labat, M.L., Nguyen, Q., Berthelet, J., Tan, X.,
26. Kawashima, Y., Kodera, Y., Singh, A., Matsumoto, M., and Matsumoto, H. Wimmer, V.C., Merino, D., Rogers, K.L., and Naik, S.H. (2021). Spatial
(2014). Efficient extraction of proteins from formalin-fixed paraffin- omics and multiplexed imaging to explore cancer biology. Nat. Methods
embedded tissues requires higher concentration of tris ( hydroxymethyl ) 18, 997–1012. https://doi.org/10.1038/s41592-021-01203-6.
aminomethane. Clin. Proteomics 11, 4. https://doi.org/10.1186/1559- 42. Merritt, C.R., Ong, G.T., Church, S.E., Barker, K., Danaher, P., Geiss, G.,
0275-11-4. Hoang, M., Jung, J., Liang, Y., McKay-Fleisch, J., et al. (2020). Multiplex
27. Meier, F., Brunner, A.D., Frank, M., Ha, A., Bludau, I., Voytik, E., Kaspar- digital spatial profiling of proteins and RNA in fixed tissue. Nat.
Schoenefeld, S., Lubeck, M., Raether, O., Bache, N., et al. (2020). Biotechnol. 38, 586–599. https://doi.org/10.1038/s41587-020-0472-9.
diaPASEF: parallel accumulation–serial fragmentation combined with 43. Zhang, B., Wang, J., Wang, X., Zhu, J., Liu, Q., Shi, Z., Chambers, M.C.,
data-independent acquisition. Nat. Methods 17, 1229–1236. https://doi. Zimmerman, L.J., Shaddox, K.F., Kim, S., et al. (2014). Proteogenomic
org/10.1038/s41592-020-00998-0. characterization of human colon and rectal cancer. Nature 513,
28. Demichev, V., Messner, C.B., Vernardis, S.I., Lilley, K.S., and Ralser, M. 382–387. https://doi.org/10.1038/nature13438.
(2020). DIA-NN: neural networks and interference correction enable 44. Derks, J., Leduc, A., Huffman, R.G., Specht, H., Ralser, M., Demichev, V.,
deep proteome coverage in high throughput. Nat. Methods 17, 41–44. and Slavov, N. (2021). Increasing the throughput of sensitive proteomics
https://doi.org/10.1038/s41592-019-0638-x. by plexDIA. Nat. Biotechnol. 41, 50–59.
29. Hammad, S., Hoehme, S., Friebel, A., Von Recklinghausen, I., Othman, A., 45. Friedrich, C., Schallenberg, S., Kirchner, M., Ziehm, M., Niquet, S., Haji,
Begher-Tibbe, B., Reif, R., Godoy, P., Johann, T., Vartak, A., et al. (2014). M., Beier, C., Neudecker, J., Klauschen, F., and Mertins, P. (2021).
Protocols for staining of bile canalicular and sinusoidal networks of human, Comprehensive micro-scaled proteome and phosphoproteome charac-
mouse and pig livers, three-dimensional reconstruction and quantification of terization of archived retrospective cancer repositories. Nat. Commun.
tissue microarchitecture by image processing and analysis. Arch. Toxicol. 12, 3576. https://doi.org/10.1038/s41467-021-23855-w.
88, 1161–1183. https://doi.org/10.1007/S00204-014-1243-5/FIGURES/9.
46. Thielert, M., Itang, C., Ammar, C., Schober, F., Bludau, I., Skowronek, P.,
30. Guo, T., Kouvonen, P., Koh, C.C., Gillet, L.C., Wolski, W.E., Röst, H.L., Wahle, M., Zeng, W.-F., Zhou, X.-X., Brunner, A.-D., et al. (2022). Robust
Rosenberger, G., Collins, B.C., Blum, L.C., Gillessen, S., et al. (2015). dimethyl-based multiplex-DIA workflow doubles single-cell proteome
Rapid mass spectrometric conversion of tissue biopsy samples into per- depth via a reference channel Author list. https://doi.org/10.15252/msb.
manent quantitative digital proteome maps. Nat. Med. 21, 407–413. 202211503.
https://doi.org/10.1038/nm.3807.
47. Perez-Riverol, Y., Csordas, A., Bai, J., Bernal-Llinares, M., Hewapathirana,
31. Coscia, F., Doll, S., Bech, J.M., Schweizer, L., Mund, A., Lengyel, E., S., Kundu, D.J., Inuganti, A., Griss, J., Mayer, G., Eisenacher, M., et al.
Lindebjerg, J., Madsen, G.I., Moreira, J.M., and Mann, M. (2020). A (2019). The PRIDE database and related tools and resources in 2019:
streamlined mass spectrometry–based proteomics workflow for large- improving support for quantification data. Nucleic Acids Res. 47, D442–
scale FFPE tissue analysis. J. Pathol. 251, 100–112. https://doi.org/10. D450. https://doi.org/10.1093/nar/gky1106.
1002/path.5420. €dler, S.C., Wallmann, G., Metousis, A., Bérouti, M.,
48. Schmacke, N.A., Ma
32. Cunningham, R.P., and Porat-Shliom, N. (2021). Liver zonation – revisiting Harz, H., Leonhardt, H., Mann, M., and Hornung, V. (2023). SPARCS, a
old questions with new technologies. Front. Physiol. 12, 1433. https://doi. platform for genome-scale CRISPR screening for spatial cellular pheno-
org/10.3389/FPHYS.2021.732929/BIBTEX. types. https://doi.org/10.1101/2023.06.01.542416.

Cell Systems 14, 1002–1014, November 15, 2023 1013


ll
Methods
49. Frankenfield, A.M., Ni, J., Ahmed, M., and Hao, L. (2022). Protein contam- comprehensive analysis of (prote)omics data. Nat. Methods 13,
inants matter: building universal protein contaminant libraries for DDA and 731–740. https://doi.org/10.1038/nmeth.3901.
DIA proteomics. J. Proteome Res. 21, 2104–2113. https://doi.org/10. 51. Cox, J., and Mann, M. (2012). 1D and 2D annotation enrichment: a statis-
1021/acs.jproteome.2c00145. tical method integrating quantitative proteomics with complementary
50. Tyanova, S., Temu, T., Sinitcyn, P., Carlson, A., Hein, M.Y., Geiger, T., high-throughput data. BMC Bioinformatics 13, S12. https://doi.org/10.
Mann, M., and Cox, J. (2016). The Perseus computational platform for 1186/1471-2105-13-S16-S12.

1014 Cell Systems 14, 1002–1014, November 15, 2023


ll
Methods

STAR+METHODS

KEY RESOURCES TABLE

REAGENT or RESOURCE SOURCE IDENTIFIER


Antibodies
Recombinant Anti-Sodium Potassium Abcam Cat#ab76020;
ATPase antibody [EP1845Y] RRID: AB_1310695
Recombinant Alexa Fluor 647 Anti-CD3D Abcam Cat#ab198937; RRID: AB_2889190
antibody [EP4426]
CD20 Monoclonal Antibody (L26), Alexa Thermo Fisher Scientific Cat# 53-0202-80, RRID: AB_10734357
Fluor 488, eBioscience
Pan Cytokeratin Monoclonal Antibody Thermo Fisher Scientific Cat# 41-9003-80, RRID: AB_11217482
(AE1/AE3), eFluor 570, eBioscience
Biological samples
Murine liver FFPE tissue from C57BL/6 mice The Jackson Laboratory https://www.jax.org/strain/000664
Human tonsil FFPE tissue Institute of Pathology at Charite https://pathologie-ccm.charite.de/
University Hospital
Chemicals, peptides, and recombinant proteins
n-Dodecyl-beta-Maltoside (DDM) Sigma-Aldrich Cat# D4641-500MG
Endoproteinase Lys-C Promega Cat# VA1170
Proteomics grade modified trypsin Promega Cat# V5117
Tris(2-carboxyethyl)phosphine Sigma-Aldrich Cat# C4706-2G
hydrochloride
Acetonitrile (ACN) HPLC-grade VWR Cat# 83640.290
Isopropanol (ISO) Sigma-Aldrich Cat# 1070222511
Triethylammonium bicarbonate pH Merck T7408-100ML
8.5 (TEAB)
Formic acid Merck Millipore Cat# 1.00264.1000
Trifluoroacetic acid Sigma-Aldrich Cat# 96924-250ML-F
2-chloroacetamide Sigma-Aldrich Cat# C0267-100G
EnVision FLEX Target Retrieval Solution Agilent Dako Cat# K8004
High pH (50X)
Microscopy Neo-Clear Sigma-Aldrich Cat# 1.09843.5000
Odyssey Blocking Buffer LI-COR Biosciences Cat# 927-70001
Prolong Diamond antifade mounting Invitrogen Cat# P36961
medium
Aqua Poly Mount Polysciences Europe GmbH Cat# 18606-20
Pepsin solution for antigen retrieval Agilent Dako Cat# S3002
Deposited data
Mass spectrometric raw files This paper ProteomeXchange: PXD042367
List of hepatocyte specific markers 10.1016/j.cmet.2014.11.002 1-s2.0-S1550413114004999-mmc5.xlsx
(Azimifar et al.22)
Software and algorithms
DIA-NN version 1.8.1 Demichev et al.28 https://github.com/vdemichev/DiaNN
R version 4.2.2 The R Project for Statistical https://www.r-project.org/
Computing
Perseus version 1.6.15.0 Tyanova et al.50 https://maxquant.net/perseus/
Leica Laser Microdissection software Leica Microsystems https://www.leica-microsystems.com/
version 8.3.0.08259 products/microscope-software/p/leica-
lmd-software/
(Continued on next page)

Cell Systems 14, 1002–1014.e1–e5, November 15, 2023 e1


ll
Methods

Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
Zeiss ZEN version 3.7 Carl Zeiss AG https://www.zeiss.com/microscopy/de/
produkte/software/zeiss-zen.html
LAS X software version 3.7.524914 Leica Microsystems https://www.leica-microsystems.com/
products/microscope-software/p/leica-
las-x-ls/
Qupath version 0.4.3 Bankhead et al.39 https://qupath.github.io/
Biological Image Analysis Single Cell Technologies https://single-cell-technologies.com/bias-
Software (BIAS) 2/BioStudies Archive accession number S-
BSST820
Bruker Compass Data Analysis Bruker Daltonik GmbH https://www.bruker.com/en/products-and-
Software version 6.0 solutions/mass-spectrometry/ms-
software.html
Qupath_to_LMD function: Contour This paper https://doi.org/10.5281/zenodo.8414787
export from Qupath to the Leica LMD7
Other
C18 Evotips (EV2013, Evotip Evosep Biosystems https://www.evosep.com/evotip/
Pure, Evosep)
96-well plate Thermo Fisher Scientific https://www.thermofisher.com/order/
catalog/product/de/en/AB1300
384-well low-binding plate Eppendorf Cat# 0030129547
Super PAP-pen liquid Science Services Cat# N71312-N
blocker mini
Cover glass Corning Cat# CLS2980223, #1.5
PPS frame slides Leica Cat# 11600294
PEN glass slides Carl Zeiss Cat# 15350731

RESOURCE AVAILABILITY

Lead contact
Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Fabian
Coscia (fabian.coscia@mdc-berlin.de).

Materials availability
This study did not generate new materials.

Data and code availability


d The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium (http://proteomecentral.
proteomexchange.org) via the PRIDE partner47 with the dataset identifier PXD042367. All data reported in this paper will be
shared by the lead contact upon request.
d The source code for processing the shapes used for laser microdissection has been deposited and is freely available at github.
com/CosciaLab/Qupath_to_LMD. The DOI is listed in the key resources table.
d Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

EXPERIMENTAL MODEL AND SUBJECT DETAILS

Mouse experiments and organ harvesting


For the mouse liver proteome experiments, 6- 8 weeks old female C57BL/6 mice from Jackson Laboratory were used. C57BL/6 mice
were housed in individually ventilated cages in a specific pathogen-free mouse facility at the Max-Delbru €ck Center for Molecular
Medicine (Berlin, Germany).
For liver excision, anesthetized mice were sacrificed by cervical dislocation, and the livers were removed, rinsed twice in ice-cold
PBS, and transferred to 4% formaldehyde solution for fixation (fixation for at least 24h to 48h). Thereafter, livers were paraffin-
embedded for further histological analyses. The animal experiments were performed in accordance with the United Kingdom Coor-
dinated Committee on Cancer Research (UKCCR) guidelines and were approved by local governmental authorities (Landesamt fu €r
Gesundheit und Soziales Berlin, Germany).

e2 Cell Systems 14, 1002–1014.e1–e5, November 15, 2023


ll
Methods

Human tissue samples


Tonsil tissues were obtained from two female patients aged 34 and 36. Both presented with recurrent chronic tonsillitis and under-
went bilateral tonsillectomy. The pathological examination revealed hypertrophy and hyperplasia of the lymphoid follicles as well as
enlarged germinal centers. Furthermore, an abundance of collagen fibers within the stroma could be observed. There were no signs
of active inflammation or malignancy.
Resected tonsils were fixed in 10% buffered formalin before gross processing. After overnight fixation, the specimens were weighed,
measured, and macroscopically evaluated. Afterward, the specimens were cut into 5-mm-thick slices. Two representative slices were
embedded in paraffin. The embedded tissue blocks were cut into 2-mm-thick sections, and stained with hematoxylin and eosin for his-
tological examination. The tissue blocks were then stored at room temperature at the archive of the Institute of Pathology at Charité Uni-
versity Hospital, Campus Mitte. The study was performed according to the ethical principles for medical research of the Declaration of
Helsinki and approval was obtained from the Ethics Committee of the Charité University Medical Department in Berlin (EA1/222/21).

METHOD DETAILS

Hematoxylin-eosin staining
Briefly, PEN glass slides (Carl Zeiss, 15350731) were treated with UV light for 1 hour. PPS frame slides (Leica, 11600294) were used
directly for the next steps. FFPE tissue sections were cut with a microtome (5 mm or 10 mm-thick), air dried at 37  C overnight and
heated at 60 C for 10 minutes to facilitate better tissue adhesion. Next, tissue sections were deparaffinized by washing
2x5 minutes in Neo Clear (Sigma Aldrich, 1.09483.5000), followed by a series of 99%, 80% and 70% ethanol for 2 minutes, respec-
tively, and rehydrated by immersing in milliQ water three times. Then, slides were stained in Mayer’s hematoxylin for three minutes
and immersed in tap water for another ten minutes, rinsed in milliQ water and stained with eosin for 30 seconds. Subsequently, the
slides were dehydrated by submerging in 70%, 80%, and 99% ethanol serially. Samples were finally air-dried and stored at RT until
imaging. Before imaging, a cover glass (Corning, CLS2980223, #1.5) was mounted with Aqua Poly Mount medium (Polysciences Eu-
rope GmbH, 18606-20).

Immunofluorescence staining
Following tissue sectioning, mounting on LMD slides and de-paraffinization (described above), two epitope retrieval methods were
compared: heat-induced (HIER) and protease (pepsin)-induced (PIER, 10 min) epitope retrieval. HIER was done by submerging in
EnVision FLEX Target Retrieval Solution High pH solution (diluted to 1X) (Agilent Dako, cat.no. K8004) and heating in a steamer at
95 C for 20 min, and subsequently cooled down in a pre-heated PBS buffer at room temperature for 30 min. Odyssey Blocking Buffer
(LI-COR BioScience, 927-70001) was used for blocking in a humidified chamber for 30 min at room temperature. PIER was done by
applying pepsin (Agilent Dako, cat.no. S3002) on the tissue slide for 5 minutes at 37 C, and immersed in PBS solution to stop the
reaction. The slides were washed two times with PBS and air dried before antibody staining.
For murine liver tissue stains, the primary antibody targeting Na/K-ATPase (stock concentration 0.563 mg/ml, dilution 1:100, Ab-
cam, ab76020) was diluted in Odyssey Blocking Buffer and incubated overnight at 4  C in a humidified chamber. Next, tissue spec-
imens were washed 3x in PBS and secondary antibodies for the visualization of Na/K-ATPase (Alexa Fluor 488 donkey anti-rabbit,
stock concentration 2 mg/ml, dilution 1:250, A32790, Invitrogen) were diluted in Odyssey Blocking Buffer and applied for 1 hour at
room temperature in the dark. After staining, slides were washed 3x in PBS, counterstained by Hoechst (dilution 1:1000 in PBS,
Thermo Fisher Scientific, #62249) for 10 minutes and followed by three washes in PBS and two washes in milliQ. Before imaging,
a cover glass (#1.5) was mounted using ProLong Diamond anti-fade mounting medium (Thermo Fisher Scientific, P36961).
For tonsil tissue stains following tissue sectioning, mounting on LMD slides and de-paraffinization, the tissues were subjected to
heat-induced epitope retrieval as described above. Odyssey Blocking Buffer was used for blocking in a humidified chamber for
30 min at room temperature. Next, conjugated primary antibodies targeting CD20 (stock concentration 0.5mg/ml, dilution 1:50, Ther-
mofisher, 53-0202-80, Alexa Fluor 488), CD3 (stock concentration 0.5 mg/ml, dilution 1:100, Abcam, ab198937, Alexa Fluor 647), and
pan-cytokeratin (stock concentration 0.2 mg/ml, dilution 1:100, Thermofisher, 41-9003-80, eFluor 570) were diluted in Odyssey
Blocking Buffer (and incubated overnight at 4  C in a humidified chamber. Tissue specimens were washed 4x in PBS, counterstained
by Hoechst (dilution 1:1000 in PBS, Thermo Fisher Scientific, 62249) for 10 minutes, washed 4x in PBS and 2x in milliQ water. Sub-
sequently, the slides were dehydrated by submerging them in 70%, 80%, and 99% ethanol serially. Before imaging, a cover glass
was mounted with ProLong Diamond anti-fade mounting medium.

High-resolution microscopy
Images of immunofluorescence-labeled tonsil tissue sections were acquired using an Axioscan 7 system (Zeiss), equipped with
wide-field optics, a Plan-A photochromat 10x/0.45 M27 objective and a quadruple-band filter set for Alexa fluorescent dyes. The
wide-field acquisition was performed using the Colibri 7 LED light source and an AxioCam 712m camera. Images were obtained auto-
matically with Zeiss ZEN 3.7 (blue edition) at non-saturating conditions (16-bit dynamic range).
Images of immunofluorescently-labeled and hematoxylin-eosin stained murine liver tissue sections were acquired on the Leica
LMD7 system using the LAS X software (version 3.7.524914, Leica Microsystems), an HC PL FLUOTAR 10x/0.32 DRY objective
and the DF7000T camera. The microscope was equipped with the following filter sets: LMD-Dapi, LMD-Cy3, LMD - Alexa594,

Cell Systems 14, 1002–1014.e1–e5, November 15, 2023 e3


ll
Methods

LMD-YFP, LMD-CY5, LMD-BGR, GFP. After imaging, the cover glass was finally removed through gentle agitation of the slide in PBS.
After one dip in milliQ water for salt removal, the slide was air-dried and stored at 4 C until laser microdissection.

Image analysis and contour export for laser microdissection


Image analysis was performed in QuPath (version 0.4.3) and BIAS (BioStudies Archive accession number S-BSST820). Annotations
of different regions of interest were manually created in QuPath after image analysis. The assignment of three reference points (x-y
coordinates) is required for precise contour transfer between the screening and laser microdissection microscopes. Contours and
reference points were exported in a geojson format and translated into the .XML format compatible with the Leica LMD7 software.
The code for processing the shapes is available at github.com/CosciaLab/Qupath_to_LMD, it uses geopandas (Version 0.12.2) and
py-lmd48 (Version 1.0.0).

Laser microdissection
We used the Leica LMD 7 system and Leica Laser Microdissection V 8.3.0.08259 software for the collection of tissue contours. De-
pending on the contour size, tissue was cut with a 20x or 63x objective in fluorescence or brightfield mode. The following laser set-
tings were used for the 20x objective (HC PL FL L 20x/0.40 CORR): power 56, aperture 1, speed 15, middle pulse count 1, final pulse
-1, head current 37 – 45% (depending on tissue type and section thickness), pulse frequency 801 and offset 101. For the 63x objec-
tive (HC PL FLUOTAR L 63x/0.70 CORR XT), the settings were: power 59, aperture 1, speed 50, middle pulse count 1, final pulse -1,
head current 45-50%, pulse frequency 3000, offset 101.
Contours were cut and sorted into a low-binding 384-well plate (Eppendorf 0030129547) configured over the ‘universal holder’
function with one empty well between samples.

Sample preparation for LC-MS analysis


A detailed sample preparation protocol is provided in the supplemental information (Methods S1). Tissue samples were collected by
manual cutting or by automated cutting after contour import into low-binding 384-well plates. For mouse liver tissue samples (5- or
10-mm-thick sections cut with a microtome), regions of 600 mm2 - 100,000 mm2 were collected. To concentrate tissue pieces at the
bottom of each well after LMD collection, 15 ml of acetonitrile was added to each well, briefly vortexed and vacuum dried (15min at
60 C). Another well inspection is recommended before proteomics sample preparation to ensure high collection efficiency.
We tested three different protocols, DDM-based, ACN-based and a combination of DDM and ACN. The lysis buffer for the DDM-
based protocol consisted of 0.1% DDM, 5mM TCEP, 20mM CAA and 0.1M TEAB in water. 2ml of lysis buffer was added to each sam-
ple well using the MANTIS Liquid Dispenser (Formulatrix, V3.3 ACC RFID, software version 4.7.5) and the high-volume diaphragm
chips (Formulatrix, cat.no. 233128). The plate was closed with a PCR ComfortLid (Hamilton), and heated at 95 C for 60 minutes.
Then, samples were shortly cooled down, and 1ml of LysC was added (prediluted in water to 2 ng/ml) and digested for minimum 2
hours at 37 C in the thermal cycler (50 C lid temperature). Subsequently, 1ml of trypsin was added (prediluted in water to 2 ng/ml)
and incubated overnight at 37  C in the thermal cycler. The next day, digestion was stopped by adding trifluoroacetic acid (TFA, final
concentration 1% v/v), and samples were vacuum dried before peptide clean-up.
For the ACN-based protocol, the lysis buffer consisted of 5mM TCEP, 20mM CAA, 0.1M TEAB diluted in water. 2 ml of lysis buffer
was added to each sample well, the plate was closed with PCR ComfortLid, and heated at 95 C for 60 minutes in a thermal cycler
(Bio-Rad S1000 with 384-well reaction module) at a constant lid temperature of 110 C. Next, 1 ul of 100% ACN was added to each
well, and the plate was incubated for an additional 60 min in a thermal cycler at 75 C at a constant lid temperature of 110 C. Then,
samples were shortly cooled down, and 1 ml of LysC was added (prediluted with 0.1M TEAB, 30% ACN water to 2 ng/ml) and digested
for 2 hours at 37 C in the thermal cycler (50 C lid temperature). Subsequently, 1 ml of trypsin was added (prediluted with 0.1M TEAB,
10% ACN water to 2 ng/ml) and incubated overnight at 37  C in the thermal cycler. The next day, digestion was stopped by adding
trifluoroacetic acid (TFA, final concentration 1% v/v), and samples were vacuum dried before peptide clean-up.
For the combined DDM/ACN-based protocol, the lysis buffer for the DDM-based protocol consisted of 0.1% DDM, 5mM TCEP,
20mM CAA and 0.1M TEAB in water. 2ml of lysis buffer was added to each sample well using the MANTIS Liquid Dispenser and the
high-volume diaphragm chips. The plate was closed with a PCR ComfortLid, and heated at 95 C for 60 minutes. Then, samples were
shortly cooled down, and 1 ml of LysC was added (2 ng/ml in 0.1M TEAB [pH 8.5] and 30% ACN in milliQ water) and digested for min-
imum 2 hours at 37 C in the thermal cycler (50 C lid temperature). Subsequently, 1 ml of trypsin was added (2 ng/ml containing 10%
ACN and 0.1M TEAB [pH 8.5] in milliQ water.) and incubated overnight at 37  C in the thermal cycler. The next day, digestion was
stopped by adding trifluoroacetic acid (TFA, final concentration 1% v/v), and samples were vacuum dried before peptide clean-up.

Peptide clean-up with C-18 tips


Evotip (Evosep, Odense, Denmark) based peptide clean-up was performed as recommended by the manufacturer. Briefly, 20 ul of
buffer B (99.9% ACN, 0.1% FA) was added to each C-18 tip (EV2013, Evotip Pure, Evosep) and centrifuged at 700 rpm for 1 minute.
Then, 20 ul of buffer A (99.9% water, 0.1% FA) was added from the top of each C-18 tip, activated in isopropanol for 20 seconds and
centrifuged again at 700 rpm for 1 minute. Digested tissue samples were then loaded onto Evotips, washed once with 20 ul buffer A
and finally eluted with 20 ul buffer B to a 96-well plate (Thermo Fisher Scientific, AB1300), and vacuum dried (15 min at 60 C).

e4 Cell Systems 14, 1002–1014.e1–e5, November 15, 2023


ll
Methods

Samples were stored at 20  C until liquid chromatography–mass spectrometry (LC–MS) analysis. For LC-MS analysis, 4.2 ml of MS
loading buffer (3% acetonitrile, 0.1% TFA in water) was added, the plate was vortexed for 10 seconds and centrifuged for 1 minute at
700g. 4 ml were finally injected into the mass spectrometer.

Liquid chromatography–mass spectrometry (LC – MS) analysis


LC-MS analysis was performed with an EASYnLC-1200 system (Thermo Fisher Scientific) connected to a trapped ion mobility spec-
trometry quadruple time-of-flight mass spectrometer (timsTOF SCP and timsTOF Pro2, Bruker Daltonik) with a nano-electrospray ion
source (CaptiveSpray, Bruker Daltonik). The autosampler was configured to pick samples from 384- and 96-well plates.
Peptides were loaded on a 20-cm home-packed HPLC column (75-mm inner diameter packed with 1.9-mm ReproSil-Pur C18-AQ
silica beads, Dr. Maisch).
Peptides were separated using a linear gradient from 7-30% buffer B (0.1% formic acid and 90% ACN in LC-MS grade water) in
14 minutes, followed by an increase to 60% for 1 minute and a 1.5-minute wash in 90% buffer B at 250 nl min-1. Buffer A consisted of
0.1% formic acid in LC-MS grade water. The total gradient length was 21 minutes. A column oven was used to keep the column tem-
perature constant at 40 C.
For dia-PASEF analysis, we used a dia-PASEF method with 8 dia-PASEF scans separated into 3 ion mobility windows per scan
covering a 400-1000 m/z range by 25 Th windows and an ion mobility range from 0.64 to 1.37 Vs cm-2. The mass spectrometer
was operated in high sensitivity mode, with an accumulation and ramp time at 100 ms, capillary voltage set to 1750V and the collision
energy as a linear ramp from 20 eV at 1/K0 = 0.6 Vs cm-2 to 59 eV at 1/K0 = 1.6 Vs cm-2. The collision energy was ramped linearly as a
function of ion mobility from 59 eV at 1/K0 = 1.6 V s cm-2 to 20 eV at 1/K0 = 0.6 V s cm-2.

Proteomics raw data analysis


We used DIA-NN28 (version 1.8.1) for dia-PASEF raw file analysis and spectral library generation.
Spectral library generation
For spectral library generation to analyze dia-PASEF data, human and mouse FASTA files were downloaded from Uniprot (2022
release, UP000000589_10090 Mus Musculus, UP000005640_9606, downloaded on April 10th and April 8th 2022 respectively).
DIA-NN in silico predicted libraries were generated by providing the human or mouse FASTA file and frequently found contaminants49
(mouse + mouse tissue contaminants, or human + universal contaminants). Deep learning-based spectra, RTs and IMs prediction
were enabled for the appropriate mass range of 300-1200 m/z. N-terminal M excision was enabled and cysteine carbamidomethy-
lation was enabled as a fixed modification. A maximum of 2 missed cleavages was allowed, and the precursor charge set to 2 - 4. For
the generation of project-specific refined libraries, in-silico-generated mouse and human libraries were used to search 20-50 raw files
of high-sample amounts, such that the optimal total ion current was reached (see Figures 2 and 3). The refined murine liver library
consisted of 68,006 precursors, 61,554 elution groups and 8,225 protein groups. The refined human tonsil library consisted of
47,999 precursors, 44,675 elution groups and 8,137 protein groups.
Search of dia-PASEF raw files with refined libraries
DIA-NN was operated in the default mode with minor adjustments. Briefly, MS1 and MS2 accuracies were set to 15.0, scan windows
to 0 (assignment by DIA-NN), isotopologues were enabled, no MBR for project-specific DIA-NN-refined libraries, heuristic protein
inference and no shared spectra. Enabling MBR for refined libraries improved data completeness of single-cell samples (Figure
S2G). No difference was observed for higher input samples. Proteins were inferred from genes, neural network classifier was set
to single-pass mode, quantification strategy as ‘Robust LC (high precision)’. Cross-run normalization was set to ‘RT-dependent’, li-
brary generation as ‘smart profiling’, speed and Ram usage as ‘optimal results’.

QUANTIFICATION AND STATISTICAL ANALYSIS

Proteomics data analysis was performed with Perseus50 (version 1.6.15.0) and within the R environment (https://www.r-project.org/,
version 4.2.2) with the following packages: ggplot2 (v3.4.2), FactoMineR (v2.8), factoextra (v 1.0.7.999), plotly (v4.10.1), reshape2
(v1.4.4), viridis (v0.6.3), UpSetR (v1.4.0). For differential expression analysis (t-test or ANOVA, Figures 3 and 4), data were filtered
to keep only proteins with 70% non-missing data in at least one group. Missing values were imputed based on a normal distribution
(width = 0.3, downshift = 1.8) before statistical testing. For multi-sample (ANOVA) or pairwise proteomic comparisons (two-sided un-
paired t-test), a permutation-based FDR of 5% was applied to correct for multiple hypothesis testing. Principal component analysis
was performed in R (see packages above). 1D pathway enrichment analysis51 (Figure 4I) was done in Perseus based on the KEGG
(https://www.genome.jp/kegg/) and Reactome Pathway Database (reactome.org), using a Benjamini-Hochberg FDR cut-off of 0.05.
The minimum category size was set to 5.

Cell Systems 14, 1002–1014.e1–e5, November 15, 2023 e5

You might also like